EP3505188A1 - Procyclische dinukleotidkonjugate zur cytokininduktion - Google Patents

Procyclische dinukleotidkonjugate zur cytokininduktion Download PDF

Info

Publication number
EP3505188A1
EP3505188A1 EP17306981.6A EP17306981A EP3505188A1 EP 3505188 A1 EP3505188 A1 EP 3505188A1 EP 17306981 A EP17306981 A EP 17306981A EP 3505188 A1 EP3505188 A1 EP 3505188A1
Authority
EP
European Patent Office
Prior art keywords
cdn
pro
formula
bam
specifier
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP17306981.6A
Other languages
English (en)
French (fr)
Inventor
Thierry Lioux
Fabienne Vernejoul
Cédric BOULARAN
Michèle TIRABY
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Invivogen SAS
Original Assignee
Invivogen SAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Invivogen SAS filed Critical Invivogen SAS
Priority to EP17306981.6A priority Critical patent/EP3505188A1/de
Priority to EP18833077.3A priority patent/EP3731874A1/de
Priority to PCT/EP2018/097128 priority patent/WO2019129880A1/en
Priority to US16/958,463 priority patent/US11730817B2/en
Publication of EP3505188A1 publication Critical patent/EP3505188A1/de
Priority to US18/226,515 priority patent/US20240165242A1/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment

Definitions

  • the present invention provides a Pro-cyclic dinucleotide (Pro-CDN) comprising a STING agonist cyclic dinucleotide which is coupled to a linker system.
  • the Pro-CDNs of the present invention can be metabolized at a targeted site into CDNs and exert their full immunomodulatory effects at said targeted site.
  • the present invention also provides conjugates wherein a Pro-CDN is conjugated to a Biologically Active Molecule (BAM) such as e.g. a cytotoxic molecule, a lipid, a protein, a peptide, a nucleic acid, a sugar or a PRR ligand.
  • BAM Biologically Active Molecule
  • the invention provides also methods related to the use of such compounds to perform their activities at their targeted sites, to exert cytotoxic, cytostatic or immunomodulatory effects, to treat or to prevent diseases such as cancers, immunological disorders or infections.
  • the Pro-CDN compound of the present invention is a compound of Formula (I): wherein:
  • the present invention concerns a Pro-CDN compound wherein a CDN compound which is a STING agonist compound is coupled to a linker system to enable the delivery of said CDN to a specific cell or tissue and thus exerts its activity to said targeted cell or tissue after being metabolized at the targeted site.
  • the Pro-CDN compound may further comprise a spacer either linked to the connector or to the specifier.
  • the spacer is a compound of Formula (IV): wherein: X 3 is -O- or -NH-, m, n and p are an integer ranging from 0 to 12 (i.e. 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12).
  • BAM-CDN conjugate is particularly interesting because it allows the specific release of the BAM and of the CDN within a specific cell/tissue.
  • the present invention also provides a BAM-CDN conjugate wherein the BAM is connected to a CDN through a linker system which is able to release both CDN and BAM so that they can exert their own activities at a targeted site.
  • the linker system possesses a number of key attributes, including the requirement to be stable in plasma after drug administration for an extended period of time such that the BAM-CDN conjugate can specifically target e.g. immune or cancer cells.
  • the BAM-CDN conjugate Upon processing, uptake or internalization, the BAM-CDN conjugate is able to release the CDN such that the CDN can bind to an intracellular target STING.
  • the linker system can have a profound effect on the physico-chemical properties of the BAM-CDN conjugate.
  • most of the CDNs are polar molecule with poor penetration into cells.
  • linking them to a BAM via a linker system containing a hydrophobic moiety can increase the uptake of CDNs.
  • the BAM-CDN conjugate comprising the Pro-CDN compound of Formula (I) and a biologically active molecule can be linked directly to the Pro-CDN compound of Formula (I) or through a spacer.
  • the BAM-CDN conjugate is a compound of the Formulae (V a ) to (V f ): wherein:
  • the present invention provides a Pro-CDN conjugated to a Biologically Active Molecule (BAM) to enhance the CDN activity and/or exert a further biological activity at a targeted site defined by the enzymatic cleavage(s) of the specifier(s).
  • BAM Biologically Active Molecule
  • the present invention provides a Pro-CDN comprising a STING ligand CDN moiety which is coupled to a linker system which can be cleaved outside, in the vicinity or in the intracellular space of a target site defined by the enzymatic cleavage of the specifier.
  • the Pro-CDN of Formula (I) comprises at least one mono- or diphosphorothioate CDN of Formula (II a ) and an enzymatically cleavable specifier, wherein the CDN and the specifier are linked together through a spontaneous self-eliminating connector of Formulae (III a ) to (III g ).
  • the specifier and the connector form a linker system.
  • the spontaneous self-eliminating connector releases the full CDN activity at the targeted site as summarized in Schemes 1.1 and 1.2.
  • the scheme 1.2 describes the release of a CDN when the connector is, for example, of Formula (III a ).
  • STING is meant to include, without limitation, homologous and/or orthologous STING molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
  • the present invention provides also a BAM-CDN conjugate comprising a Biologically Active Molecule linked to a Pro-CDN, notably via a spacer.
  • the spacer is connected either to a specifier or to a connector of the linker system of the Pro-CDN.
  • the BAM-CDN conjugate is selectively activable outside, in the vicinity or in the intracellular space of a target site defined by the enzymatic cleavage(s) of the specifier(s).
  • the BAM-CDN conjugate of Formulae (V a ) to (V f ) of this invention comprises at least one mono- or diphosphorothioate CDN of Formula (II a ), a linker system comprising a specifier linked to a connector of Formulae (III a ) to (III g ) and a BAM as defined above via notably a spacer.
  • the specifier is cleaved by a specific enzyme, the spontaneous self-eliminating connector releases the active form of CDN and the BAM linked to specifier or spacer-specifier at the targeted site as summarized, for example, for Formula (V a ) in Scheme 1.3.
  • the BAM-CDN conjugates of Formulae may contain another linker system between the BAM and the Pro-CDN to release the active form of CDN and the native form of BAM.
  • One aspect of the present invention provides a CDN conjugate which is selectively activable in the intracellular space.
  • Another aspect of the invention provides a cell specific CDN conjugate which is a highly selective substrate for drug-activating enzymatic cleavage of a target cell.
  • the specific release of the CDN and the BAM at the vicinity or in the intracellular space of a target site is defined by the enzymatic cleavage of the specifier.
  • specifier used herein is defined as an enzymatically cleavable unit, i.e. a substrate which can be cleaved by an enzyme, and that can thus be specifically cleaved in a specific environment depending on the presence or not of said enzyme.
  • the specifier thus allows for the specific release of the CDN and of the BAM within a specific cell/tissue depending on the specific conditions found in this specific cell/tissue.
  • cleavable specifiers There are two types of cleavable specifiers: a specific peptide sequence or a specific sugar:
  • One aspect of the present invention provides BAM-CDN conjugates which are stable in biological fluids until they reach their target.
  • the administration of the compounds according to the present invention is less toxic than the administration of the CDN or of the BAM alone; the activities of the present compounds being maximal after the specifier cleavage.
  • the specifier is typically a substrate that is specifically cleaved by an enzyme present in the vicinity of, or inside the targeted cells. More preferably, the specifier is a substrate that is specifically cleaved by an enzyme present at elevated levels in the vicinity of, or inside the target cells as compared to other parts of the body, and most preferably the enzyme is present only in the vicinity of or inside the target cells.
  • the specifier is a targeting moiety able to target specific cells and deliver a compound to the targeted cells.
  • the specifier is a substrate of a lysosomal enzyme. According to this embodiment, the specifier allows targeting the intracellular space of targeted cells/tissues....
  • the specifier is a di-, tri- or oligopeptide consisting of an amino acid sequence specifically recognized and cleaved by a protease.
  • the specifier may be chosen to be preferably targeted by a non-mammalian protease or by an endogenous mammalian protease present in the vicinity of or inside the target cells and chosen from the list (but not restricted): beta-site APP-cleaving enzyme 1 (BACE1), Cathepsin D (CTSD), Calpain-1 (CAPN1), Caspase 1 (CASP1), Caspase 2 (CASP2), Caspase 3 (CASP3), Caspase 5 (CASP5), Caspase 6 (CASP6), Caspase 7 (CASP7), Caspase 8 (CASP8), Caspase 9 (CASP9), Cathepsin B (CTSB), Cathepsin K (CTSK), Cathepsin L (CTSL), Cathepsin
  • the specifier may contain protective groups. Such compounds comprising protected specifier may not, when contacted with for instance specific enzymes, release the leaving groups. However, when deprotected and suitably activated such compounds will release leaving groups and thus such compounds comprising a protected specifier also fall under the scope of this invention.
  • specifier peptide sequence may be chosen among those which can be cleaved upon acidic condition, by an extracellular enzyme or by an intracellular enzyme.
  • the specifier sequence is selected from D-alanylphenylalanyllysine, D-valylleucyllysine, D-alanylleucyllysine, D-valylphenylalanyllysine, D-valyltryptophanyllysine D-alanyltryptophanyllysine, valylalanine, valylcitrilline, Gly-Phe-Leu-Gly, Ala-Leu-Ala-Leu; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys-Ala
  • the specifier is a dipeptide selected from the group consisting of valine-citrulline, valine-alanine.
  • Dipeptides can have lower hydrophobicity compared to longer peptides.
  • the specifier is an amino-terminal capped peptide covalently linked via the C-terminus to the self-eliminating spacer or linker system.
  • the specifier can be cleaved by a specific enzyme of tumoral tissue or tumor micro-environment.
  • the activating enzyme generates the release of the CDN and the BAM in the vicinity or inside the tumor cell.
  • proteolytic enzymes have been shown to be associated with tumor invasion and metastasis.
  • proteases like cathepsins, proteases from the urokinase-type plasminogen activator (u-PA) system or the Serine protease plasmin are all involved in tumor metastasis.
  • the proteolytically active form of plasmin is formed from its inactive Pro-enzyme form plasminogen by u-PA.
  • the tumor-associated presence of plasmin can be exploited for targeting of plasmin-cleavable conjugates or prodrugs.
  • the specifier is a substrate of cathepsins.
  • Such specifiers would thus be selected from the group consisting of valine-citrulline and valine-alanine.
  • the specifier can be a sugar such as galactose.
  • the ⁇ -galactosidase enzyme is mainly active intracellularly, it will promote the releases of CDN and BAM within the targeted cells.
  • the specifier can be a sugar such as Glucuronic acid.
  • Glucuronic acid As ⁇ -glucuronidase is highly concentrated in the microenvironment of a wide range of solid tumors including lung, breast, and gastrointestinal tract carcinomas, molecules containing Glucuronide specifier can be prepared with the aim to deliver CDN and BAM units in the vicinity of the tumor.
  • the specifier is a nitro group that can be reduced under hypoxic conditions or by nitroreductases.
  • the reactive moiety in specifier is reacted with a nucleophilic group on a targeting moiety, e.g., a thiol group, an amino group, or an aldehyde group, to form a new specifier that contains a targeting moiety.
  • a targeting moiety e.g., a thiol group, an amino group, or an aldehyde group
  • the reactive moiety of specifier is reacted with a nucleophilic group on a BAM, e.g., a thiol group, an amino group, or an aldehyde group, to form a new specifier that contains BAM as the targeting moiety.
  • a nucleophilic group on a BAM e.g., a thiol group, an amino group, or an aldehyde group
  • the technology of this invention relates to novel linker systems that can be either coupled to the CDN to form a Pro-CDN or that can be inserted between a CDN and a BAM to release each moiety at their targeted site.
  • Biologically active molecules BAM.
  • one aspect of the present invention requires conjugating a BAM to a CDN through a specifier and a self-eliminating group (connector) and optionally a spacer to target an appropriate site.
  • Targeting is defined by the specifier and after the enzymatic cleavage reaction will remove the specifier moiety from the CDN or/and BAM conjugate and selectively release the different agents in pharmacologically active form at the target site.
  • the BAM according to the present invention is e.g. selected from the group consisting of:
  • said Biologically Active Molecule is a protein.
  • the "protein” may be an antibody, antibody fragments, immunoglobulin, peptides, enzymes, growth factors, cytokines, chemokines, transcription factors, toxins, antigen peptides, hormones, carrier proteins, channels, motor function proteins, receptors, signaling proteins, scaffolding proteins, storage proteins, membrane proteins, transmembrane proteins, internal proteins, external proteins, secretory proteins, viral proteins, glycoproteins, glycopeptides, cleaved proteins, protein complexes, chemically modified proteins.
  • the Biologically Active Molecule unit is an antibody.
  • antibodies used for cancer application are Abciximab, Adalimumab, Alemtuzumab, Atlizumab, Basiliximab, Belimumab, Bevacizumab, Bretuximab vedotin, Canakinumab, Cetuximab, Ceertolizumab pegol, Daclizumab, Denosumab, Eculizumab, Efalizumab, Gemtuzumab, Golimumab, Golimumab, Ibritumomab tiuxetan, Infliximab, Ipilimumab, Muromonab-CD3, Natalizumab, Ofatumumab, Omalizumab, Palivizumab, Panitumuab, Ranibizumab, Rituximab, Tocilizumab, Tositumomab and
  • BAM is an immune-stimulatory compound.
  • said Biologically Active Molecule compound is a damage-associated molecular pattern molecules (DAMPs) or a Pathogen-Associated Molecular Pattern molecules, (PAMPs) to provide multi-PRR ligands.
  • DAMPs damage-associated molecular pattern molecules
  • PAMPs Pathogen-Associated Molecular Pattern molecules
  • BAM-CDN conjugates can be recognized by receptors of the innate immune system, such as Toll-like receptors (TLRs), Nod-like receptors, C-type lectins, cytosolic dsDNA sensors, RIG-I-like receptors and proteins involved in those pathways. These receptors can be transmembrane or intra-endosomal proteins which can prime activation of the immune system in response to infectious agents such as pathogens.
  • said immune-stimulatory compound is a toll-like receptor agonist, STING agonist, or RIG-I agonist.
  • said Biologically Active Molecule compound is a TLR1 agonist, a TLR2 agonist, a TLR3 agonist, a TLR4 agonist, a TLR5 agonist, a TLR6 agonist, a TLR7 agonist, a TLR8 agonist, a TLR9 agonist or a TLR10 agonist.
  • said Biologically Active Molecule compound is a bioactive molecule including (but not restricted) inhibitor (for example growth factors inhibitor, IDO inhibitor, MAPK/ERK inhibitors, COX inhibitor, inflammatory inhibitor, TBK1 inhibitor, angiogenesis inhibitor, Carbonic Anhydrase IX/XII Inhibitor, enzymes inhibitor used in the treatment of auto-immune, fungal, bacterial, viral and parasite diseases or in cancer treatment), agonist or a partial agonist (for example Adora-2a receptor agonist).
  • inhibitor for example growth factors inhibitor, IDO inhibitor, MAPK/ERK inhibitors, COX inhibitor, inflammatory inhibitor, TBK1 inhibitor, angiogenesis inhibitor, Carbonic Anhydrase IX/XII Inhibitor, enzymes inhibitor used in the treatment of auto-immune, fungal, bacterial, viral and parasite diseases or in cancer treatment
  • agonist or a partial agonist for example Adora-2a receptor agonist
  • said Biologically Active Molecule compound is a particle, for example (but not restricted) polymeric micelle nanoparticles, polymer coated iron oxide nanoparticles, carbon nanoparticles.
  • said Biologically Active Molecule compound is a lipid.
  • lipids may be fatty acids, glycerolipids, glycerophospholipids, sphingolipids, saccharolipids, and polyketides sterol lipids and prenol lipids.
  • the Biologically Active Molecule unit comprises a lipid to form liposomal structure with CDN or a nanoparticle to protect and to increase the uptake of CDN.
  • a liposome being a lipid vesicle is capable of containing many substances irrespective of whether such substances are water-soluble or hydrophobic and thus is expected to be a prospective carrier especially for a drug delivery system.
  • said Biologically Active Molecule compound is a fluorescent probe belonging (but not restricted) to Xanthene derivatives (fluorescein, rhodamine, Oregon green, eosin, Texas red%), Cyanine derivatives (cyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine, merocyanine%), Squaraine derivatives and ring-substituted squaraines, Naphthalene derivatives (dansyl, prodan derivatives), Coumarin derivatives (6,8-difluoro-7-hydroxycoumarin-3-carboxylic acid: pacific blue,...), oxadiazole derivatives (pyridyloxazole, nitrobenzoxadiazole, benzoxadiazole%), Anthracene derivatives (anthraquinones, including DRAQ5, DRAQ7, CyTRAK Orange), Pyrene derivatives (cascade blue, ...), Oxazine
  • said Biologically Active Molecule compound is a cytotoxic agent for example but not restricted) Actinomycin, Doxifluridine, Methotrexate, All-trans retinoic acid, Doxorubicin, Mitoxantrone, Azacytidine, Epirubicin, Oxaliplatin, Azathioprine, Epothilone, Paclitaxel, Bleomycin, Etoposide, Pemetrexed, Bortezomib, Fluorouracil, Teniposide, Carboplatin, Gemcitabine, Tioguanine, Capecitabine, Hydroxyurea, Topotecan, Cisplatin, Idarubicin, Valrubicin, Chlorambucil, Imatinib, Vemurafenib, Cyclophosphamide, Irinotecan, Vinblastine, Cytarabine, Mechlorethamine, Vincristine, Daunorubicin, Mercaptopurine, Vindesine, Docet
  • the Biologically Active Molecule compound is a radio-sensitizing molecule including (but not restricted to) nicotinamide, carbogen, metronidazole and metronidazole analogs, hypoxic cell cytotoxic agents (mitomycin-c), membrane active agents (chlorpromazine), radiosensitizing nucleosides (5-FU, Bromodeoxyuridine, Gemcitabine), texaphyrins.
  • said Biologically Active Molecule compound is the folic acid (FA).
  • the FA is one of the most widely used in anticancer nanomedicine. Folate receptor is generally overexpressed in most types of tumor cells (such as ovarian and breast cancer) and has limited expression in normal cells. Furthermore, FA has other advantages, such as non-immunogenicity, high stability and good tumor penetration. For these reasons, FA has been widely used as a tumor-targeting ligand in cancer imaging and therapy (ref 10.1016/j.xphs.2017.02.019) because of its commercial availability, small size and its easy conjugation.
  • the BAM-CDN conjugates induce cell death.
  • BAM is a protein
  • Pro-CDN units are conjugated to 1 BAM unit through connectors and/or spacer in order to increase the number of CDN bound per BAM.
  • a number of recent publications have described the use of branched linkers in combination with antibody-containing prodrugs or bioconjugates with the aim of increasing the number of drugs bound per antibody.
  • the BAM is selected from the group consisting of:
  • the present invention relates the use of a connector, such as Para-Amino-Benzyl (PAB) or Para-amino-benzyl-carbonyl (PABC) which is a self-eliminating group through 1,6-elimination principle ( Carl et al., J. Med. Chem., 1981, vol. 24, 479-480 .) and is able to release spontaneously the BAM moiety or CDN as described in Scheme 1.4.
  • a connector such as Para-Amino-Benzyl (PAB) or Para-amino-benzyl-carbonyl (PABC) which is a self-eliminating group through 1,6-elimination principle ( Carl et al., J. Med. Chem., 1981, vol. 24, 479-480 .) and is able to release spontaneously the BAM moiety or CDN as described in Scheme 1.4.
  • PAB Para-Amino-Benzyl
  • PABC Para-amino-benzyl-carbony
  • the present invention relates to the use of a connector, such as Para-Amino-Benzyl (PAB), which is able to make a phosphorothioate triester link with a CDN or such as Para-amino-benzyl-carbonyl (PABC) which is able to make a carbamate link with the BAM.
  • a connector such as Para-Amino-Benzyl (PAB) which is able to make a phosphorothioate triester link with a CDN or such as Para-amino-benzyl-carbonyl (PABC) which is able to make a carbamate link with the BAM.
  • PAB Para-Amino-Benzyl
  • PABC Para-amino-benzyl-carbonyl
  • the connector is a compound of Formulae (III a ) to (III g ):
  • the connectors of formulae (III e ) and (III f ) are a di or tri-functional chemical moiety that is capable of covalently linking to the BAM or to the CDN to promote the release the native form of the BAM or the CDN.
  • the leaving group PAB or PABC must react with functionality at the end of the specifier.
  • the end of the specifier is a carboxylic group, but it can also be another functionality according the specifier chosen.
  • a connector described above can be coupled to the BAM in order to promote the release of the native form of the BAM.
  • a hydrophilic spacer can be incorporated in the Pro-CDN or between the Pro-CDN and the BAM in order to improve hydrophilic property of the Pro-CDN or the BAM-CDN conjugate (and thus its water solubility) and spatially separate BAM from CDN. Thus, its presence may facilitate enzymatic cleavage and so enhance the kinetics of drug release.
  • the spacer can e.g. be linked directly to the specifier or to the connector.
  • the spacer is a polyamine including: polyethylenimine, polylysine (PLL), spermine, spermidine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
  • PLL polylysine
  • spermine spermine
  • spermidine pseudopeptide-polyamine
  • peptidomimetic polyamine dendrimer polyamine
  • arginine amidine
  • protamine cationic lipid
  • cationic porphyrin quaternary salt of a polyamine
  • quaternary salt of a polyamine or an alpha helical peptide.
  • the spacer is a polyethylene glycol (PEG).
  • the spacer is a compound of Formula (IV): wherein: X 3 is -O- or -NH-, m, n and p are an integer ranging from 0 to 12 (i.e. 0, 1, 2, 3,4, 5, 6, 7 ,8, 9, 10, 11 or 12).
  • the CDN unit of the present invention is a compound of Formula (II a ), including its pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs, and more specifically is a CDN with at least one phosphorothioate linkage: wherein:
  • X 2 and Y 2 are H.
  • X 2 is H and Y 2 is F.
  • X 2 is F and Y 2 is H.
  • X 2 and Y 2 are F.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (3',3') CDNs of Formula (II b ), including its pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs: wherein X 1 , Y 1 , X 2 , Y 2 , B 1 , B 2 , and R 1 are as defined above.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (2',3') CDNs of Formula (II c ) and CDNs of Formula (II d ), including their pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs: wherein X 1 , Y 1 , X 2 , Y 2 , B 1 , B 2 , R 1 and R 2 are as defined above.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (3',2') CDNs of Formula (II e ) and CDNs of Formula (II f ), including their pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs: wherein X 1 , Y 1 , X 2 , Y 2 , B 1 , B 2 , R 1 and R 2 are as defined above.
  • the presence of Fluoride (F) or hydrogen (H) atoms at the 2' or 3' position of the sugar is indispensable for the esterification of the phosphorothioate.
  • F Fluoride
  • H hydrogen
  • the esterification reaction provides a large scale of acyclic dinucleotide.
  • the preferred sugar residues of the nucleoside are pentofuranosyl sugars selected from the group consisting of D-enantiomers of ribose or xylose and their modified derivatives on position 2' Formula (VIII a ) or 3' Formula (VIII b ): wherein X is F or H.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (3',3') CDNs of Formulae (II g ), including their pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs, with X 1 and X 2 are fluoride atom, Y1 and Y2 are hydrogen atom: wherein B 1 , B 2 , and R 1 are as defined above.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (2',3') CDNs of Formula (II h ) and CDNs of Formula (II i ) including their pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs, with Y 1 and X 2 are fluoride atom, X 1 and Y 2 are hydrogen atom: wherein B 1 , B 2 , R 1 and R 2 are as defined above.
  • the CDN of compounds of Formula (II a ) is a monophosphorothioate molecule and can be selected from (3',2') CDNs of Formula (II j ) and CDNs of Formula (II k ), including their pharmaceutically acceptable salts, esters, stereoisomers, tautomers, solvates or prodrugs, with Y 2 and X 1 are fluoride atom, X 2 and Y 1 are hydrogen atom: wherein B 1 , B 2 , R 1 and R 2 are as defined above.
  • Compounds bearing a phospho or phosphorothioate diester group have a negatively charged ionic nature at physiological pH.
  • the therapeutic activity of such compounds is consequently limited, on account of the low diffusion of negatively charged compounds across biological lipid membranes.
  • charged compounds do not diffuse efficiently across cell membranes, or indeed across the cerebral barrier.
  • the coupling to a cleavable Linker system via the phosphorothioate diester link of compound of Formulae (II a ) to (II k ) is one solution to drug delivery and/or bioavailability issues.
  • the phosphorothioate modification acts as a prodrug by masking the polar functional group according to previously published data ( Gong-Xin et al: « Chapter 3.6. Prodrugs of Phosphonates, Phosphinates and Phosphates » Prodrugs Challenges and Reward Part1. Springer New York, US, vol.5.1, 2007, p. 923-964 ).
  • the mission of such prodrug was to mask the sulfur atoms of phosphorothioate groups in CDN analogues so that they are neutral at physiological pH and hence have a better uptake into cells.
  • the phosphorothioate moiety is a mandatory element to couple the CDN to the connector.
  • the Pro-CDN can be synthetized as described in scheme 2.1:
  • the Specifier-Connector of Formula (XII) can be connected to the STING agonist of Formula (II a ) by a direct esterification of a phosphorothioate diester link between two nucleosides of the CDN.
  • the reaction can be realized in a mix of water and acetone to reach the compound of Formula (XIII) which can be deprotected to give the Pro-CDN of Formula (I b ).
  • W 2 is link between of the specifier and the connector the nature of W 2 depends of X 7 and X 8 functions and is described below.
  • W 1 can be an amide, an ester, a urea, a disulfide bridge, a carbamate, a hydrazone, an imine, an oxime, a triazole group;
  • the synthesis of the conjugate BAM-Pro-CDN requires the preparation of compound of Formula (XIX), then the coupling of this compound with the CDN of Formula (II a ) can provide the desired conjugate of Formula (V g ) by esterification of phosphorothioate link as defined above.
  • P 1 , BAM, Specifier, X 3 , X 4 , X 5 , X 6 , X 1 , Y 1 , X 2 , Y 2 , Z 1 , R 1 , B 1 , B 2 , X 7 , X 8 , W 2 and W 3 are as defined above.
  • W 3 is link between of the BAM and the connector the nature of W 3 depends of X 8 and X 3 function.
  • W 3 can be an amide, an ester, a disulfide bridge.
  • W 2 can be:
  • Forming amides is one of the most important reactions for conjugation between a Biologically Active Molecule of Formulae (VII a ) or (XXXII) and the compound of Formulae (XVI) or (XXX).
  • amines can react with carboxyl acid via some familiar activating reagents, such as N- hydroxysuccinimide (NHS), 2-Succinimido-1,1,3,3-tetra-methyluronium tetrafluoroborate (TSTU), and Benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate (PyBOP).
  • NHS N- hydroxysuccinimide
  • TSTU 2-Succinimido-1,1,3,3-tetra-methyluronium tetrafluoroborate
  • PyBOP Benzotriazol-1-yloxytripyrrolidinophosphonium hexafluorophosphate
  • amines of lysines are commonly used for linking the acid terminal of spacer of compound of Formula (I) because lysines are usually exposed on the surface of the antibodies and therefore are easily accessible.
  • acid functions of glutamic acid (Glu) or Aspartic acid (Asp) can be used for linking the amine terminal of spacer of compound of Formula (I).
  • alcohol derivatives can react with carboxylic acid derivatives with known method.
  • Urea or thiourea can be obtained from isocyanate or isothiocyanate derivative and an amine derivative.
  • thiols for example, interchain cysteine residues in monoclonal antibodies or thiol of the Biologically Active Molecule, as attachment sites for drug molecules is one of the most used conjugation methods.
  • disulfide bonds can be used as potential conjugation sites.
  • the disulfide bonds can be reduced by tris(2-carboxyethyl) phosphine (TCEP) or dithiothreitol (DTT), which results in thiol groups that are available for conjugating with CDN-linker molecules.
  • TCEP tris(2-carboxyethyl) phosphine
  • DTT dithiothreitol
  • cysteine residues can be modified through addition of thiols to electrophiles such as maleimides.
  • the conjugate could be achieved by reducing the disulfide bonds of the Biologically Active Molecule and then adding to maleimides. Addition to maleimides is the most common method for attaching drugs to antibodies.
  • Adcetris® which was approved by the FDA for the treatment of patients with Hodgkin's lymphoma after failed autologous stem cell transplantation or patients with systemic anaplastic large-cell lymphoma after the failure of at least one prior multi-agent chemotherapy regimen, was produced by this method in which a maleimide-functionalized drug was conjugated to the interchain cysteine residues of an anti-CD30 antibody.
  • Maleimide-based antibody-drug conjugates were recently found to have limited stability in blood circulation, which would lower the efficacy of the conjugates and damage healthy tissue.
  • This reagent comprises a 3-arylpropionitrile (APN) group that replaces the maleimide and allows for the preparation of remarkably stable conjugates.
  • APN 3-arylpropionitrile
  • Amines of the Biologically Active Molecule could react with the hydroxyls that derived from the linkers in the effect of phosgene, 4-nitrophenyl chloroformate, etc . and form the carbamate containing CDN-linkers.
  • alcohols can react with chloroformates to form carbonates.
  • chloroformates For example, Moon et al. conjugated 7-ethyl-10-hydroxycamptothecin (SN-38) derivatives to hMN-14, a humanized anti-CEACAM5 mAb, via a carbonate bond.
  • Conjugation via aldehydes is another method for linking CDN-linker to a Biologically Active Molecule with an amine, a hydrazine or a hydroxylamine function.
  • Azides can react with alkynes to form triazoles through click chemistries, such as copper-catalyzed azide-alkyne cycloaddition (CuAAC).
  • click chemistries such as copper-catalyzed azide-alkyne cycloaddition (CuAAC).
  • This approach can be used when an azide or an alkyne function is present or can be introduced on the Linker or on the BAM moeity.
  • the azide function present on the BAM could react with a dibenzylcyclooctyne (DBCO) containing on CDN-linker without any catalytic agent.
  • DBCO dibenzylcyclooctyne
  • Stimulator of interferon genes can act as a cytosolic DNA sensor wherein cytosolic DNA and unique bacterial nucleic acids called cyclic dinucleotides (CDN) are recognized by STING, and therefore STING agonists.
  • CDN cyclic dinucleotides
  • cyclic dinucleotide represents a class of cyclic molecules with one phosphodiester linkage and one phosphorothioate diester linkage, between two nucleosides. This includes (3',5')-(3',5') nucleotide linkages (abbreviated as (3',3')); (3',5')-(2',5') nucleotide linkages (abbreviated as (3',2')); (2',5')-(3',5') nucleotide linkages (abbreviated as (2',3')).
  • Pro-CDN refers to a CDN coupled to a linker system.
  • the Pro-CDNs of the present invention need to be metabolized at their targeted sites into CDNs to exert their full immunomodulatory effects.
  • linker system refers to a connector linked to a specifier.
  • connector refers to a spontaneous self-eliminating group linking a molecule (CDN or BAM) to the specifier.
  • reporter refers to an enzymatically cleavable unit.
  • BAM Biologically active molecule refers to any moiety that can be coupled to a Pro-CDN via notably a spacer.
  • spacer refers to a hydrophilic group.
  • nucleoside refers to a glycosylamine comprising a nitrogenous base and a five-carbon sugar, wherein the nitrogenous base is bound to the five-carbon sugar via a beta-glycosidic linkage.
  • nucleotide refers to any nucleoside linked to a phosphate group at position 5', 3' or 2' of the sugar moiety.
  • “Pharmaceutically acceptable salts” include those salts derived from pharmaceutically acceptable inorganic or organic bases. Suitable salts include for instance those derived from alkali metals such as potassium and sodium or from alkaline earth metals such as calcium and magnesium or from organic bases such as triethylamine. Due to the negatively charged ionic nature of the phospho or phosphorothioate diester linkage, the counter-ion forming the salt is positively charged.
  • Steps include in particular enantiomers.
  • the compounds of the invention may be enantiomerically pure or provided as a mixture of enantiomers.
  • the tautomers of the compounds of the invention include compounds deriving from tautomerisation of a hypoxanthine moiety or a guanine moiety as follows:
  • solvate denotes a compound formed by solvation, for example as a combination of solvent molecules with molecules or ions of a solute.
  • Well known solvent molecules include water, alcohols and other polar organic solvents. Alcohols include methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol, and t-butanol. Alcohols also include polymerized alcohols such as polyalkylene glycols (e.g., polyethylene glycol, polypropylene glycol).
  • the best-known and preferred solvent is typically water, and solvate compounds formed by solvation with water are termed hydrates.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a Pro-CDN or a BAM-CDN conjugate of the invention and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is an immunogenic composition or vaccine adjuvant comprising a Pro-CDN or a BAM-CDN conjugate of the invention.
  • the pharmaceutical composition is a composition comprising a Pro-CDN or a BAM-CDN conjugate of the invention and one or more immunostimulatory agents.
  • the pharmaceutical composition is a vaccine or immunogenic composition comprising an antigen or antigen composition and a Pro-CDN or a BAM-CDN conjugate of the invention.
  • the pharmaceutical composition may comprise conventional excipients in conventional proportions, with or without additional active compounds or principles, and the unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • the excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipient thereof.
  • the compound of the invention is administered in a pharmaceutically effective amount.
  • the amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient, vehicle or carrier.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Such unit doses may therefore be administered once or more than once a day.
  • Such pharmaceutical compositions may be prepared by any of the methods well known in the pharmacy art.
  • compositions may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual), rectal, inhaled, intranasal, topical (including buccal, sublingual or transdermal), vaginal or injectable (including subcutaneous, intramuscular, parenteral, intravenous or intradermal) route.
  • Such compositions may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier(s) or excipient(s).
  • Another object of the present invention is Pro-CDN or a BAM-CDN conjugate of the invention for use in a therapeutic treatment in humans or animals.
  • the Pro-CDN or a BAM-CDN conjugate of the invention may be used as therapeutic agents in a monotherapy or in a combination therapy, such as chemoimmunotherapy.
  • therapeutic agent refers to one or more substances that are administered to a human or animal in order to achieve some kind of therapeutic effect in that human or animal, including to prevent, cure, or mitigate the effects of, infection or disease, and/or to otherwise improve the health of that human or animal.
  • the term "monotherapy” refers to the use of a single substance and/or strategy to treat a human or animal in any clinical or medical context, as opposed to the use of multiple substances and/or strategies to treat a human or animal in the same clinical or medical context, regardless of whether the multiple substances and/or strategies are used sequentially in any order or concurrently.
  • One object of the invention is the Pro-CDN or a BAM-CDN conjugate of the invention for use in the treatment of a disease that may be alleviated by the induction of an immune response via the STING pathway.
  • One particular aspect of the invention is a Pro-CDN or a BAM-CDN conjugate of the invention for use as an immunomodulatory agent.
  • the Pro-CDN or a BAM-CDN conjugate of the invention can be administered as immunotherapy to a human or an animal to induce in vivo production of one or more cytokines that are therapeutically beneficial to that human or animal.
  • This type of immunotherapy could be used alone or in combination with other treatment strategies, whether sequentially in any order, or concurrently. It could be used to prevent, cure, and/or mitigate the effects of, infection or disease in that human or animal, and/or to modulate the immune system of that human or animal to achieve some other therapeutic benefit.
  • Immunotherapy refers to any medical treatment in which one or more components of a human's or animal's immune system is deliberately modulated in order to directly or indirectly achieve some therapeutic benefit, including systemic and/or local effects, and preventative and/or curative effects.
  • Immunotherapy can involve administering one or more immune agents (see definition above), either alone or in any combination, to a human or animal subject by any route ( e.g. orally, intravenously, dermally, by injection, by inhalation, etc.), whether systemically, locally or both.
  • Immunotherapy can involve provoking, increasing, decreasing, halting, preventing, blocking or otherwise modulating the production of cytokines, and/or activating or deactivating cytokines or immune cells, and/or modulating the levels of immune cells, and/or delivering one or more therapeutic or diagnostic substances to a particular location in the body or to a particular type of cell or tissue, and/or destroying particular cells or tissue. Immunotherapy can be used to achieve local effects, systemic effects or a combination of both.
  • immunosens refers to the ensemble, or to any one or more components, of the molecules, substances (e.g. bodily fluids), anatomic structures (e.g. cells, tissue and organs) and physiologic processes involved in preventing infection in the body, in protecting the body during infection or during disease, and/or in helping the body to recuperate after infection or disease.
  • substances e.g. bodily fluids
  • anatomic structures e.g. cells, tissue and organs
  • physiologic processes involved in preventing infection in the body, in protecting the body during infection or during disease, and/or in helping the body to recuperate after infection or disease.
  • immune agent refers to any endogenous or exogenous substance that can interact with any one or more components of the immune system.
  • immunodeficiency virus includes antibodies, antigens, vaccines and their constituent components, nucleic acids, synthetic drugs, natural or synthetic organic compounds, cytokines, natural or modified cells, synthetic analogs thereof, and/or fragments thereof.
  • immunosuppressed describes the state of any human or animal subject whose immune system is functionally diminished, deactivated or otherwise compromised, or in whom one or more immune components is functionally diminished, deactivated or otherwise compromised. "Immunosuppression” can be the cause, consequence or byproduct of disease, infection, exhaustion, malnutrition, medical treatment or some other physiologic or clinical state.
  • immunomodulating substance refers to any substance that, upon administration to a human or animal, directly influences the functioning of the immune system of that human or animal.
  • immunomodulators include, but are not limited to, antigens, antibodies and small-molecule drugs.
  • the Pro-CDN or a BAM-CDN conjugate of the invention are used for cytokine induction as immunotherapy of immunosuppressed individuals.
  • the present invention thus discloses a method for inducing cytokine in immunosuppressed individuals, said method comprising administering to a patient in need thereof a Pro-CDN or a BAM-CDN conjugate of the invention.
  • Another aspect of the invention is a Pro-CDN or a BAM-CDN conjugate of the invention for use as immunoadjuvant.
  • the Pro-CDN or a BAM-CDN conjugate of the invention can be used for cytokine induction immunotherapy as vaccine adjuvant therapy.
  • a Pro-CDN or a BAM-CDN conjugate of the invention would be administered to a human or animal subject that has received, is receiving or will receive a vaccination.
  • the benefits provided by the present invention might include enhanced efficacy of the vaccination against the target antigen, reduced toxicity of the vaccination, reduced adverse side effects of the vaccination, or enhanced immune protection of the human or animal subject.
  • vaccine refers to a biological preparation administered to a human or animal in order to elicit or enhance a specific immune system response and/or protection against one or more antigens in that human or animal.
  • vaccination refers to treatment of a human or animal with a vaccine or to the act of administering a vaccine to a human or animal.
  • adjuvant refers to a secondary therapeutic substance that is administered together (either sequentially in any order, or concurrently) with a primary therapeutic substance to achieve some kind of complimentary, synergic or otherwise beneficial effect that could not be achieved through use of the primary therapeutic substance alone.
  • An adjuvant can be used together with a vaccine, chemotherapy, or some other therapeutic substance.
  • Adjuvants can enhance the efficacy of the primary therapeutic substance, reduce the toxicity or side effects of the primary therapeutic substance, or provide some kind of protection to the subject that receives the primary therapeutic substance, such as, but not limited to, improved functioning of the immune system.
  • Another object of the present invention is a Pro-CDN or a BAM-CDN conjugate of the invention for use in the treatment of cancer or pre-cancerous syndromes, bacterial infection or infectious diseases, such as viral infection, in particular an AIDS infection or an HIV infection.
  • cancer refers to the physiological condition in subjects that is characterized by unregulated or dysregulated cell growth or death.
  • cancer includes solid tumors and blood-born tumors, whether malignant or benign.
  • the cancer is acinar adenocarcinoma, acinar carcinoma, acral-lentiginous melanoma, actinic keratosis, adenocarcinoma, adenocystic carcinoma, adenosquamous carcinoma, adnexal carcinoma, adrenal rest tumor, adrenocortical carcinoma, aldosterone secreting carcinoma, alveolar soft part sarcoma, amelanotic melanoma, ameloblastic thyroid carcinoma, angiosarcoma, apocrine carcinoma, Askin's tumor, astrocytoma, basal cell carcinoma, basaloid carcinoma, basosquamous cell carcinoma, biliary cancer, bone cancer, bone marrow cancer, botryoid sarcoma, brain cancer, breast cancer, bronchioalveolar carcinoma, bronchogenic adenocarcinoma, bronchogenic carcinoma, carcinoma ex pleomorphic adenoma, cervical
  • the cancer is chosen from the following group: melanoma, prostate cancer, hepatocarcinoma, colon cancer, bladder cancer, breast cancer, cholangiocellular cancer, leukemia, lung cancer, lymphoma, nasopharyngeal cancer, ovarian cancer, pancreatic cancer and urothelial cancer.
  • the cancer is from the following group: melanoma, lymphoma colon and pancreatic cancer.
  • the present invention thus discloses a method for treating a cancer or pre-cancerous syndromes, a bacterial infection or infectious diseases, such as viral infection, in particular an AIDS infection or an HIV infection, said method comprising administering to a patient in need thereof a Pro-CDN or a BAM-CDN conjugate of the invention.
  • Another object of the invention is a therapeutic combination comprising a Pro-CDN or a BAM-CDN conjugate of the invention and a therapeutic agent.
  • Another object of the invention is a kit-of-parts comprising a Pro-CDN or a BAM-CDN conjugate of the invention and a chemotherapeutic agent, for use in the treatment of cancer.
  • kit-of-parts it is meant a combined preparation wherein the active ingredients are combined together or physically separated for use in a combined therapy by simultaneous administration or sequential administration to the patient.
  • the Pro-CDN or a BAM-CDN conjugate of the present invention can be used for cytokine induction immunotherapy in combination with chemotherapy.
  • a Pro-CDN or a BAM-CDN conjugate of the invention would be administered together with one or more chemotherapeutic agents, sequentially in any order or concomitantly, to a cancer patient to stop the growth of, shrink and/or destroy tumors in that patient.
  • the chemoimmunotherapy resulting from the combination of cytokine induction, Provided by the compound(s) of the present invention, and cytotoxicity, provided by the chemotherapeutic agent(s), might be less toxic to the patient, cause fewer side effects in the patient and/or exhibit greater antitumor efficacy than would the chemotherapeutic agent(s) when used as monotherapy.
  • chemotherapeutic agent refers to one or more chemical substances that are administered to a human or animal in order to kill tumors, or slow or stop the growth of tumors, and/or slow or stop the division of cancerous cells and/or prevent or slow metastasis. Chemotherapeutic agents are often administered to treat cancer, but are also indicated for other diseases.
  • chemotherapy refers to medical treatment of a human or animal with one or more chemotherapeutic agents (see definition above).
  • chemoimmunotherapy refers to the combined use, whether sequentially in any order or concurrently, of chemotherapy substances and/or strategies, and immunotherapy substances and/or strategies. Chemoimmunotherapy is often employed to treat cancer, but can also be employed to treat other diseases.
  • the present invention thus discloses a method for treating cancer, said method comprising administering to a patient in need thereof:
  • the kit-of-parts can contain both compounds in a single pharmaceutical composition, such as a tablet, or in separate pharmaceutical compositions. When the compounds are not administered simultaneously, the kit-of-parts will contain each compound in separate pharmaceutical compositions either in a single package or in separate pharmaceutical compositions in separate packages.
  • the kit-of-parts can also be provided by instruction, such as dosage and administration instructions.
  • dosage and administration instructions can be of the kind that are provided to a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, for example by a drug product label, or they can be of the kind that are provided by a doctor, such as instructions to a patient.
  • such sequential administration may be close in time or remote in time.
  • administration of the other agent several minutes to several dozen minutes after the administration of the first agent, and administration of the other agent several hours to several days after the administration of the first agent are included, wherein the lapse of time is not limited, For example, one agent may be administered once a day, and the other agent may be administered 2 or 3 times a day, or one agent may be administered once a week, and the other agent may be administered once a day and the like.
  • a Pro-CDN or a BAM-CDN conjugate of the invention would be administered to an immunosuppressed human or animal subject to induce in vivo production of one or more cytokines that directly or indirectly enhance the immune system of that human or animal.
  • Subjects that might benefit from such treatment include those suffering from autoimmune disorders, immune system deficiencies or defects, microbial or viral infections, infectious diseases, or cancer.
  • Example 1 shows preparative methods for synthesizing the conjugates
  • Example 2 shows methods for the biological evaluation of these conjugates.
  • Anhydrous solvents and reagents suitable for nucleoside and nucleotide synthesis were purchased and were handled under dry argon or nitrogen using anhydrous technique.
  • Amidite coupling reactions and cyclizations were performed in anhydrous acetonitrile or pyridine under dry argon or nitrogen.
  • the starting materials for all reactions in dry pyridine were dried by concentration (three times) from pyridine.
  • Preparative silica-gel flash chromatography was performed using Fluka 60 ⁇ high-purity grade or Merck Grade 9385 silica using gradients of methanol in dichloromethane.
  • Analytical LC/ES MS was performed on an Agilent 1290 Infinity UHPLC system coupled to a diode array detector (DAD) Agilent 1260 Infinity and an Agilent 6130 Quadrupole mass spectrometer equipped with an electrospray ionization source (ESI) and controlled by Chemstation software.
  • the LC system was equipped with an Aquity CSH C18 50x2.1 mm 1.7 ⁇ m column using gradients of 10 mM ammonioum formate and acetonitrile at 300 ⁇ l/min flow.
  • the UV detection wavelength was 254 nm.
  • the mass spectrometer was operated in positive and negative ESI modes
  • Preparative HPLC was performed on a Waters preparative 150Q HPLC system monitoring at 254 nm on a SunFire Prep C18 5 ⁇ m OBD 30 x 150mm column using gradients of 10 mM ammonium formate and acetonitrile at a flow rate 60 mL/min.
  • the 1 H NMR spectra were acquired on either a Bruker 300 MHz (Fourrier 300) at room temperature and reported in ppm downfield.
  • Molecular sieves (MS) 3 ⁇ were employed after drying the commercially supplied product at 250 °C for 12 h under vacuum.
  • the commercial nucleoside phosphoramidites were supplied from Chemgenes.
  • a 150 mL peptide reactor was purged with nitrogen and then charged with 2 g of 2-CTC resin and 80 mL of DCM the mixture was stirred for 15 min and the solvent was drained this was repeated 2 times, then the resin was suspended in 80 mL of DMF.
  • the resin-DMF mixture was stirred at rt for 30 min.
  • Fmoc-AlaOH (2.90 g, 9.30 mmol) in 40 mL DMF
  • DIEA (162 mL, 9.30 mmol
  • the mixture containing the Fmoc-AlaOH was charged to the reactor with the resin and stirred. After 4 h the reactor was drained. Active sites on the resin were end-capped with a mixture of DIEA:MeOH (1:9 mL). This mixture was then stirred at rt for 1 h. The bed was drained, washed with 2 times with 40 mL DMF, 2 times with 40 mL DCM, and one time with 40 mL DMF. The last wash demonstrated a negative UV test.
  • the dipeptide Fmoc-Val-Ala-2CTC was prepared using a similar procedure to that described for Intermediate 2.3 with FmocAlaOH (0.682 g, 2.19 mmol) and FmocValOH (0.743 g, 2019 mmol). Then the Fmoc protecting group was cleaved by addition of 60 mL of 20% piperidine in DMF under stirring at rt for 30 min. The reactor was drained and then the treatement was repeated one time. The resin bed was then washed with 3 times with 40 mL DMF, 3 times with 40 mL of DCM, 3 times with 40 mL of MeOH and 3 times with 40 mL of DMF. The last wash was then sampled for piperidine levels by qualitative ninhydrin test.
  • Palmitic acid 0.562 g, 2.19 mmol
  • DIEA 0.381 mL, 2.19 mmol
  • HATU 0.83 g, 2.19 mmol
  • the mixture was stirred at 10°C to dissolve solid.
  • the cooled solution was charged to the solid phase reactor.
  • the flask was then washed with 5 mL of DMF and the wash charged to the SPPS reactor.
  • the mixture was stirred at rt for 4 h.
  • the resin beads were then sampled for the completion of the reaction by a Keiser Test.
  • the reactor was drained and the resin bed washed with 4 times with 20 mL DMF, 3 times with 20 mL of DCM, 4 times with 20 mL of MeOH, 3 times with 20 mL of DMF and 8 times with 20 mL of DCM.
  • the Intermediate 2.8 was obtained from Intermediate 2.7 (0.215 g, 0.504 mmol) using a similar procedure to that described for Intermediate 2.4 to provide 0.14 g (54 % yield) of Intermediate 2.8.
  • the Intermediate 2.10 was obtained from Intermediate 2.9 (0.14 g, 0.263 mmol) using a similar procedure to that described for Intermediate 2.6 to provide 0.14 g (99 % yield) of Intermediate 2.10 .
  • Compound 4 belongs to Formula (V a ) and comprises a monophosphorothioate CDN (CL797), a PAB moiety as a connector, a Val-Ala as a specifier and a BAM consisting of a saturated fatty acid (C 16 carbon chain) to facilitate CDN penetration into cell.
  • the immunomodulatory activity of these CDNs analogs modified with linker systems was ascertained in in vitro -cell based assay and in live mammalian cells.
  • These compounds induced the production of multiple cytokines specifically the production of Type I interferons and/or pro-inflammatory cytokines, as indirectly determined by an ISG54 (interferon-stimulated gene) reporter assay (Fensterl et al., 2008).
  • Example 2.1 Evaluation of Pro-CDN for their ability to activate STING-dependent cytokine induction in vitro in a human or murine reporter cell line
  • the in vitro STING agonist activity disclosed in the present invention has been measured by monitoring of the IRF pathway.
  • the IRF pathway has been investigated by using the two following ISG reporter cell lines obtained directly from InvivoGen. They are described here and provided with their corresponding InvivoGen catalog code.
  • RAW - LuciaTM ISG (InvivoGen catalog code: rawl-isg): These cells were generated from the RAW 264.7 murine macrophage cell line (ATCC® TIB-71TM). They enable study of IRF signaling pathway, by assessing the activity of a secreted luciferase (Lucia), measured in cell culture supernatant by using QUANTI-LucTM (InvivoGen; catalog code: rep-qlc1), a luminometric enzyme assay that measures luciferase expression.
  • QUANTI-LucTM InvivoGen; catalog code: rep-qlc1
  • THP1-DualTM (InvivoGen catalog code: thpd-nfis): These cells were derived from the human monocytic cell line THP-1 by stable integration of two inducible reporter constructs. They enable simultaneous study of two signaling pathways: the NF- ⁇ B pathway, by monitoring the activity of secreted embryonic alkaline phosphatase (SEAP); and the IRF pathway, by assessing the activity of a secreted luciferase (Lucia).
  • SEAP secreted embryonic alkaline phosphatase
  • IRF pathway by assessing the activity of a secreted luciferase (Lucia).
  • Both reporter proteins can be readily measured in the cell culture supernatant by using QUANTI-BlueTM (InvivoGen catalog code: rep-qb1), a SEAP detection reagent that turns purple/blue in the presence of SEAP (quantified by measuring the optical density from 620 nm to 655 nm), and QUANTI-LucTM (InvivoGen; catalog code: rep-qlc1), a luminometric enzyme assay that measures luciferase expression to report on ISG54 expression (as an indicator of IFN- ⁇ / ⁇ production).
  • Figure 1 illustrates the results from this experiment.
  • Figures 1A and 1D illustrate the fold induction for tested compound (CL793, CL802 and CL804) relative to untreated cells and compared to the reference compound (2'3'-cGAMP and unmodified CDN CL702 or CL797) when used at the same concentration in an ISG murine reporter cell line.
  • Figures 1B and 1E depict the ISG response in a human reporter cell line while Figures 1C and 1F the STING-dependent NF- ⁇ B response.
  • the different dose response curves show that each one of the read out for the tested CDNs (CL793, CL802 or CL804) induces a similar to a greater ISG or NF- ⁇ B response than does 2'3'-cGAMP, the endogenous STING agonist in mammals.
  • the modified Pro-CDN compounds display the same activity than the unmodified CDN CL702 or CL797. This finding demonstrated that modification of STING ligands to promote their binding to any biologically active molecules are able to keep on their ability to induce ISG and NF- ⁇ B pathways.
  • Example 2.2 Evaluation of Pro-CDNs for their ability to induce cytokines ex vivo in whole blood from healthy human donors
  • Type I IFNs HEK-BlueTM IFN- ⁇ / ⁇ -KO-STING: These cells, in which the STING gene has been inactivated, are derived from HEK293 cell line known as HEK-BlueTM IFN- ⁇ / ⁇ (InvivoGen catalog code: hkb-ifnab). HEK-BlueTM IFN- ⁇ / ⁇ cells enable detection of bioactive human type I IFNs through monitoring of activation of the ISG3 pathway. These cells were generated by stable transfection of HEK293 cells with the human STAT2 and IRF9 genes to obtain a fully active Type-I IFN signaling pathway. The other genes of the pathway (IFNAR1, IFNAR2, JAK1, TyK2 and STAT1) are naturally expressed in sufficient amounts.
  • IFNAR1, IFNAR2, JAK1, TyK2 and STAT1 are naturally expressed in sufficient amounts.
  • the cells were further transfected with a SEAP reporter gene under control of an IFN ⁇ / ⁇ -inducible ISG54 promoter.
  • This promoter comprises five IFN-stimulated response elements (ISREs) fused to a minimal promoter of the human ISG54 gene, which is unresponsive to activators of the NF- ⁇ B or AP-1 pathways.
  • ISREs IFN-stimulated response elements
  • Stimulation of HEK-BlueTM IFN- ⁇ / ⁇ cells with human IFN- ⁇ or IFN- ⁇ activates the JAK/STAT/ISGF3 pathway and subsequently induces production of SEAP. Production of type I IFNs in these cells is measured using QUANTI-BlueTM.
  • Each blood sample was diluted (1:2 [v/v]) in RPMI medium and aliquoted into 96-well plates (180- ⁇ L wells) containing either CDN ( ⁇ 10-fold serial dilution starting from 50 ⁇ M). The plates were incubated at 37 °C in a 5% CO 2 incubator for 18 hours. Then, the supernatants were collected, transferred into the corresponding wells of round-bottom 96-well plates, and either stored at -80 °C, or immediately tested in the appropriate reporter cell line.
  • a new 96-wells plate was prepared for each reporter cell lines tested, as follows: 10 ⁇ L of supernatant from the previous plate were added to the corresponding well in the new reporter cell plate. Then, a 190- ⁇ L aliquot of cells of the desired reporter cell line, previously harvested in medium containing heat-inactivated serum and counted, was added to each well (approximately 50,000 cells/well), and the plate was incubated for approximately 20 hours. The desired cytokine induction activity was determined using the QUANTI-BlueTM assay, as previously described. Briefly, 20 ⁇ L of supernatant from the previously incubated plate was transferred to the corresponding well of a new 96-well plate in which 180 ⁇ L of QUANTI-BlueTM reagent had previously been added.
  • Results presented in Figure 2 summarizes the induction of type I IFNs for CDN-linker system compounds (CL793, CL802 and CL804) relative to the reference compound (2'3'-cGAMP) or unmodified CDN (CL702 or CL797 respectively) in the whole-blood assay for each compound in this assay.
  • the tested Pro-CDN CL793 provides, like the unmodified corresponding CDN (CL702), superior induction of type I interferons compared to the reference compound 2'3'-cGAMP.
  • the dose-response curve in Figure 2B shows that the tested Pro-CDN CL804 induce a similar type I IFNs production to the one induced by the unmodified corresponding CDN, CL797.
  • Example 2.3 Evaluation of BAM-CDN conjugates for their ability to activate STING-dependent cytokine induction in vitro in a human or murine reporter cell line
  • Figure 3 illustrates the results from this experiment.
  • Figure 4A illustrates the fold induction for tested compounds (CL794 and CL808) relative to untreated cells and compared to the reference compound (2'3'-cGAMP and unmodified corresponding CDN CL797) when used at the same concentration in an ISG murine reporter cell line.
  • Figure 4B shows the ISG response in a human reporter cell line while Figure 3C the STING-dependent NF- ⁇ B response.
  • the different dose response curves show that the BAM-CDN CL808 induces an ISG or NF- ⁇ B response to a greater extent than does 2'3'-cGAMP, the endogenous STING agonist in mammals.
  • the BAM-CDN compound CL808 displays a much higher activity compared to the unmodified corresponding CDN, CL797. It has to be noted that we observed toxicity at higher concentration of CL808 for both cell lines. This finding demonstrates that the coupling of STING ligands to a biologically active molecules does not modify their ability to induce ISG and NF- ⁇ B pathways.
  • BAM is a lipid moiety that is able to form liposomal structure with CDN or a nanoparticle to protect and to increase the uptake of CDN. The strong gain in activity compare to unmodified CDN could be explained by a better penetration of the CDN to reach its intracellular target.
  • Example 2.4 Evaluation of BAM-CDNs for their ability to induce cytokines ex vivo in whole blood from healthy human donors
  • Type I IFNs HEK-BlueTM IFN- ⁇ / ⁇ -KO-STING: These cells, in which the STING gene has been inactivated, are derived from HEK293 cell line known as HEK-BlueTM IFN- ⁇ / ⁇ (InvivoGen catalog code: hkb-ifnab). HEK-BlueTM IFN- ⁇ / ⁇ cells enable detection of bioactive human type I IFNs through monitoring of activation of the ISG3 pathway. These cells were generated by stable transfection of HEK293 cells with the human STAT2 and IRF9 genes to obtain a fully active Type-I IFN signaling pathway. The other genes of the pathway (IFNAR1, IFNAR2, JAK1, TyK2 and STAT1) are naturally expressed in sufficient amounts.
  • IFNAR1, IFNAR2, JAK1, TyK2 and STAT1 are naturally expressed in sufficient amounts.
  • the cells were further transfected with a SEAP reporter gene under control of an IFN- ⁇ / ⁇ -inducible ISG54 promoter.
  • This promoter comprises five IFN-stimulated response elements (ISREs) fused to a minimal promoter of the human ISG54 gene, which is unresponsive to activators of the NF- ⁇ B or AP-1 pathways.
  • ISREs IFN-stimulated response elements
  • Stimulation of HEK-BlueTM IFN- ⁇ / ⁇ cells with human IFN- ⁇ or IFN- ⁇ activates the JAK/STAT/ISGF3 pathway and subsequently induces production of SEAP. Production of type I IFNs in these cells is measured using QUANTI-BlueTM.
  • HEK-BlueTM IL-1R (InvivoGen catalog code: hkb-il1r): The HEK293 cell line known as HEK-BlueTM IL-1R was designed to detect bioactive human and murine IL-1 through monitoring of activation of the NF- ⁇ B and AP-1 pathways. Additionally, these cells detect bioactive IL-1 from cynomolgus monkeys, dogs, hamsters and rats. In fact, HEK-BlueTM IL-1R cells can detect IL-1 ⁇ and IL-1 ⁇ , as these cytokines bind to the same receptor, IL-1R.
  • HEK-BlueTM IL-1 ⁇ cells InvivoGen catalog code: hkb-il1b, in which the TNF- ⁇ response is blocked. Therefore, HEK-BlueTM IL-1R cells respond specifically to IL-1. These cells endogenously express the human IL-1 receptor and were stably transfected with the murine IL-1 receptor, rendering them sensitive to both human and murine IL-1 ⁇ .
  • HEK-BlueTM IL-1R cells express a SEAP reporter gene under control of an IFN- ⁇ minimal promoter fused to five NF- B and five AP-1 binding sites.
  • Binding of IL-1 ⁇ to IL-1R on the surface of HEK-BlueTM IL-1R cells triggers a signaling cascade that leads to the activation of NF- ⁇ B and subsequent production of SEAP. Production of IL-1 ⁇ in these cells is measured using QUANTI BlueTM.
  • IL6 HEK-BlueTM IL-6 (InvivoGen catalog code: hkb-hil6): HEK-BlueTM IL-6 cells allow the detection of bioactive human IL-6 by monitoring the activation of the STAT-3 pathway. These cells were generated by stable transfection of HEK293 cells with the human IL-6R gene and a STAT3-inducible SEAP reporter gene. Upon IL-6 stimulation, HEK-BlueTM IL-6 cells trigger the activation of STAT3 and the subsequent secretion of SEAP. Levels of STAT3-induced SEAP can be readily monitored using QUANTI-BlueTM.
  • HEK-BlueTM TNF- ⁇ (InvivoGen catalog code: hkb-tnfdmyd): HEK-BlueTM TNF- ⁇ cells are a HEK293 cell line that enables detection of bioactive human and murine TNF- ⁇ through monitoring of activation of the NF- ⁇ B pathway. These cells were generated by stable transfection of HEK293 cells with a SEAP reporter gene under control of an IFN- ⁇ minimal promoter fused to five NF- ⁇ B and five AP-1 binding sites. They were further rendered unresponsive to IL-1 ⁇ by knocking out the MyD88 gene. Stimulation of HEK-BlueTM TNF- ⁇ cells with TNF- ⁇ triggers activation of the NF- ⁇ B-inducible promoter and production of SEAP. Production of TNF- ⁇ in these cells is measured using QUANTI-BlueTM.
  • the ex vivo activity of BAM-CDN conjugates has been measured by monitoring the production of type I IFNs, and pro-inflammatory cytokines IL1 ⁇ / ⁇ , IL6 and TNF ⁇ in whole blood assays as described above in Example 2.2.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP17306981.6A 2017-12-29 2017-12-29 Procyclische dinukleotidkonjugate zur cytokininduktion Withdrawn EP3505188A1 (de)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP17306981.6A EP3505188A1 (de) 2017-12-29 2017-12-29 Procyclische dinukleotidkonjugate zur cytokininduktion
EP18833077.3A EP3731874A1 (de) 2017-12-29 2018-12-28 Procyclische dinukleotide und procyclische dinukleotidkonjugate zur cytokinduktion
PCT/EP2018/097128 WO2019129880A1 (en) 2017-12-29 2018-12-28 Pro-cyclic dinucleotides and pro-cyclic dinucleotide conjugates for cytokine induction
US16/958,463 US11730817B2 (en) 2017-12-29 2018-12-28 Pro-cyclic dinucleotides and pro-cyclic dinucleotide conjugates for cytokine induction
US18/226,515 US20240165242A1 (en) 2017-12-29 2023-07-26 Pro-cyclic dinucleotides and pro-cyclic dinucleotide conjugates for cytokine induction

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP17306981.6A EP3505188A1 (de) 2017-12-29 2017-12-29 Procyclische dinukleotidkonjugate zur cytokininduktion

Publications (1)

Publication Number Publication Date
EP3505188A1 true EP3505188A1 (de) 2019-07-03

Family

ID=61027402

Family Applications (2)

Application Number Title Priority Date Filing Date
EP17306981.6A Withdrawn EP3505188A1 (de) 2017-12-29 2017-12-29 Procyclische dinukleotidkonjugate zur cytokininduktion
EP18833077.3A Pending EP3731874A1 (de) 2017-12-29 2018-12-28 Procyclische dinukleotide und procyclische dinukleotidkonjugate zur cytokinduktion

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP18833077.3A Pending EP3731874A1 (de) 2017-12-29 2018-12-28 Procyclische dinukleotide und procyclische dinukleotidkonjugate zur cytokinduktion

Country Status (3)

Country Link
US (2) US11730817B2 (de)
EP (2) EP3505188A1 (de)
WO (1) WO2019129880A1 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021216572A1 (en) * 2020-04-20 2021-10-28 Massachusetts Institute Of Technology Lipid compositions for delivery of sting agonist compounds and uses thereof

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019123340A1 (en) 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
CA3084582A1 (en) 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
CA3093888A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
TWI818007B (zh) 2018-04-06 2023-10-11 捷克科學院有機化學與生物化學研究所 2'3'-環二核苷酸
CA3147890A1 (en) * 2019-07-19 2021-01-28 Immunesensor Therapeutics, Inc. Antibody-sting agonist conjugates and their use in immunotherapy
EP4134098A4 (de) 2020-04-10 2024-05-15 Ono Pharmaceutical Co Verfahren zur krebstherapie
CN111592570B (zh) * 2020-05-15 2022-04-29 清华大学 新型sting激动剂及其制备方法和应用
CN111909223A (zh) * 2020-07-17 2020-11-10 清华大学 环二核苷酸共价修饰物及其制备方法和应用
PE20231947A1 (es) * 2020-11-09 2023-12-05 Takeda Pharmaceuticals Co Conjugados de anticuerpo y farmaco
CN114569627A (zh) * 2020-12-02 2022-06-03 杭州星鳌生物科技有限公司 新型sting激动剂复合物的制备、组成及其在抗肿瘤药物中的应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998013059A1 (en) 1996-09-27 1998-04-02 Bristol-Myers Squibb Company Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
WO2016096577A1 (en) * 2014-12-16 2016-06-23 Invivogen Combined use of a chemotherapeutic agent and a cyclic dinucleotide for cancer treatment
WO2017019896A1 (en) * 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to pd-1
WO2017100305A2 (en) * 2015-12-07 2017-06-15 Opi Vi - Ip Holdco Llc Composition of antibody construct-agonist conjugates and methods of use thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JOP20170192A1 (ar) 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co داي نوكليوتيد حلقي
US10265778B2 (en) * 2017-01-16 2019-04-23 Black & Decker Inc. Accessories for oscillating power tools
AR113224A1 (es) * 2017-04-28 2020-02-19 Novartis Ag Conjugados de anticuerpo que comprenden un agonista de sting

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998013059A1 (en) 1996-09-27 1998-04-02 Bristol-Myers Squibb Company Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
WO2016096577A1 (en) * 2014-12-16 2016-06-23 Invivogen Combined use of a chemotherapeutic agent and a cyclic dinucleotide for cancer treatment
WO2017019896A1 (en) * 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to pd-1
WO2017100305A2 (en) * 2015-12-07 2017-06-15 Opi Vi - Ip Holdco Llc Composition of antibody construct-agonist conjugates and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CARL ET AL., J. MED. CHEM., vol. 24, 1981, pages 479 - 480
GONG-XIN ET AL.: "Prodrugs Challenges and Reward Parti", vol. 5.1, 2007, SPRINGER, article "Chapter 3.6. Prodrugs of Phosphonates, Phosphinates and Phosphates", pages: 923 - 964

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021216572A1 (en) * 2020-04-20 2021-10-28 Massachusetts Institute Of Technology Lipid compositions for delivery of sting agonist compounds and uses thereof

Also Published As

Publication number Publication date
EP3731874A1 (de) 2020-11-04
WO2019129880A1 (en) 2019-07-04
US20220339287A9 (en) 2022-10-27
US20240165242A1 (en) 2024-05-23
US20230116913A2 (en) 2023-04-13
US11730817B2 (en) 2023-08-22
US20210353760A1 (en) 2021-11-18

Similar Documents

Publication Publication Date Title
EP3505188A1 (de) Procyclische dinukleotidkonjugate zur cytokininduktion
KR102459468B1 (ko) 결합 링커, 그러한 결합 링커를 함유하는 세포 결합 분자-약물 결합체, 링커를 갖는 그러한 결합체의 제조 및 사용
CA3013412C (en) Specific conjugation linkers, specific immunoconjugates thereof, methods of making and uses such conjugates thereof
ES2740907T3 (es) Enlazador de sulfamida, conjugados de los mismos y métodos de preparación
KR20230074285A (ko) 비스-링키지를 사용한 세포독성 약물의 접합
Lelle et al. Octreotide-mediated tumor-targeted drug delivery via a cleavable doxorubicin–peptide conjugate
AU2021362997A1 (en) Specific conjugation of an antibody
KR20200097295A (ko) 분지형 링커를 갖는 튜불리신 유사체의 접합체
CN110167599B (zh) 包含引入β-半乳糖苷的自我牺牲型连接基团的化合物
CN113423430A (zh) 含支链连接子的鹅膏毒素偶联物
BR112019022445A2 (pt) Composição de conjugado ligante-fármaco, formulação, método para inibir a multiplicação de uma célula tumoral ou célula cancerosa ou causar apoptose em uma célula tumoral ou cancerosa, e, composto de conector de fármaco
TW202140075A (zh) 具有含肽之連接子的位置特異性抗體-藥物共軛體
ES2853582T3 (es) Moduladores del sistema inmune y composiciones
US20120094892A1 (en) Prodrugs
EP4194010A1 (de) Konjugat aus einem funktionellen arzneimittel und einem toll-like-rezeptor-7- oder -8-agonisten mit temporär inaktivierter wirkstoffstelle und verwendung davon
LOPEZ RIVAS Synthesis of integrin-targeting pro-drugs for the selective release of anti-tumor agents
TW202317196A (zh) 用於藥物遞送應用之基於兩親性胜肽的自組裝奈米顆粒
KR20220017378A (ko) 활성화 부위가 일시적으로 비활성화된 톨-유사 수용체 7 또는 8 작용자와 기능성 약물의 결합체 및 그 용도
RAPOSO MOREIRA DIAS Synthesis of RGD Peptidomimetic-Drug Conjugates for the Tumor-Targeted Delivery of Cytotoxic Agents
JP2024510927A (ja) 抗体-薬物コンジュゲートのための分岐リンカー及びその使用方法
CA3201005A1 (en) Glycoside dual-cleavage linkers for antibody-drug conjugates
CN117916253A (zh) 用于疾病治疗的甲胎蛋白生物缀合物

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20200104